118
Views
10
CrossRef citations to date
0
Altmetric
Review

The arsenal of pathogens and antivirulence therapeutic strategies for disarming them

&
Pages 1795-1806 | Published online: 27 May 2016

Abstract

Pathogens deploy an arsenal of virulence factors (VFs) to establish themselves within their infectious niche. The discovery of antimicrobial compounds and their development into therapeutics has made a monumental impact on human and microbial populations. Although humans have used antimicrobials for medicinal and agricultural purposes, microorganism populations have developed and shared resistance mechanisms to persevere in the face of classical antimicrobials. However, a positive substitute is antivirulence therapy; antivirulence therapeutics prevent or interrupt an infection by counteracting a pathogen’s VFs. Their application can reduce the use of broad-spectrum antimicrobials and dampen the frequency with which resistant strains emerge. Here, we summarize the contribution of VFs to various acute and chronic infections. In correspondence with this, we provide an overview of the research and development of antivirulence strategies.

The ascent and collapse of conventional antimicrobials

In 1929, Alexander Fleming described a method for isolating Bacillus influenzae using penicillium mold broth filtrate, which he termed penicillin.Citation1 In his report, Fleming emphasized penicillin’s use in the bacteriological laboratory and briefly mentioned its potential implications for medicine. Ironically, in that first report, Fleming recommended taking advantage of the innate penicillin resistance of gram-negative bacteria to promote their isolation and identification.Citation1 This basic scientific discovery heralded in the golden age of antimicrobials.

Although penicillin launched the age of antimicrobials, prontosil, a sulfonamide discovered by Gerhard Domagk working for IG Farben (Bayer) in 1935, was the first antimicrobial manufactured for therapeutic application.Citation2,Citation3 Neisseria and Streptococcus infections were among the first to be widely treated with sulfonamides.Citation2,Citation4,Citation5 Penicillin was not available for clinical use until the 1940s, when Ernst Chain and Howard Florey developed a method allowing for mass production.Citation6

The abundant use of antibiotics that followed in hospitals and agricultural industry initiated the surfacing of antimicrobial-resistant strains. For example, sulfonamide-resistant Neisseria gonorrhoeae isolates became wide spread in the clinical setting shortly after World War II.Citation5 Penicillin replaced sulfonamides for treatment, leading to the emergence of penicillin resistance within the next 15–20 years.Citation7Citation9 Gradually, N. gonorrhoeae developed resistance to the vast majority of other classes of antibiotics, including aminoglycosides, tetracycline, macrolides, and most recently fluoroquinolones.Citation10 In 2007, the United States Centers for Disease Control and Prevention announced cephalosporins as the only remaining option for treating N. gonorrhoeae.Citation11 Strains with reduced susceptibility to the cephalosporins cefixime and ceftriax-one have since emerged.Citation11 Over the past few decades, only one new antimicrobial class was discovered.Citation12 The next generation of antimicrobials and therapeutic strategies must withstand the force of microbial evolution; unless, we wish to confront another cohort of “super-bugs”.

Antivirulence therapies (AVTs) that target virulence factors (VFs) or their production/regulation constitute an alternative approach to classical antimicrobials. This is in contrast to current cidal or static antimicrobials that disrupt pathogen growth, thereby selecting for mutations that would allow resistant populations to take over.Citation13 The aim of many AVTs is to tax bacterial metabolism to a point that pathogens are incapable of paying the “fitness cost” associated with a prospective infectious niche. In addition, commensal microbes are unlikely to produce the targets of AVTs, thus reducing the collateral damage imposed to the host’s microbiota by current antimicrobials. However, AVTs require a greater level of forethought in their design and understanding of microbial pathogenesis, which of course come with a greater fiscal burden. In certain cases, effective implementation of AVTs requires rapid diagnostics, which is far from being a new priority in medical research. In this review, we focus on microbial VFs and the latest progress in developing AVTs against them.

A battle of the ages: common virulence traits and antivirulence strategies aimed to combat them

Establishing residency through adherence

Upon introduction to a niche, pathogens are faced with the challenge of remaining “on location” long enough to establish infection. Microorganisms can be swept away by shear forces applied from the movement of biological fluids and host cell cilia. Pathogens can use various extracellular structures for mediating adherence to host cells. Enteropathogenic Escherichia coli (EPEC) rely on bundle-forming pili (BFP) and EspA filaments for microcolony formation and early attachment to intestinal epithelial cells.Citation14Citation16 Volunteers, who ingested wild-type EPEC, exhibited frequent bouts of diarrhea; in contrast, their luckier counterparts, who ingested bfp mutant strains, had negligible symptoms.Citation17 EPEC, enterohemorragic E. coli (EHEC), and their murine model counterpart Citrobacter rodentium can form attaching and effacing lesions during infection. These pathogens use type 3 secretion systems (T3SSs) to inject the intimin receptor (Tir) into the host cell.Citation18Citation20 Translocated Tir becomes embedded into the host cell and serves as a bacterial docking mechanism via its interaction with the bacterial intimin protein.Citation18Citation20

Extraintestinal pathogenic E. coli adhere to host cell niches via the use of pili that assemble by the chaperone usher pathway (CUP) system.Citation21Citation27 The proteins adorning the tips of CUP pili are specialized adhesins with stereochemical specificity to distinct moieties depending on the CUP pilus. For example, the adhesin of type 1 pili (Fim), FimH, mediates adherence to the bladder epithelial cells by binding with glycoproteins on the host cell’s surface.Citation23,Citation28 The pyelonephritis-associated pili (pap) operon codes for the P pili components; P pili are capped by the PapG adhesin, which binds specifically to the α-d-Galp-(1→4)-β-d-Galp disaccharide moieties and mediates kidney colonization.Citation29Citation35

Antiadherence strategies to prevent bacterial colonization

Strategies aiming to prevent bacterial adherence can target the regulatory systems governing adhesin expression, disrupt the secretion of adhesins and their assembly, or block the binding between the bacterial adhesin and host receptor. In EPEC, a vaccine against BFP is in development, inspired by production of antibodies to the BfpA component, in children naturally infected with EPEC.Citation36Citation38 Research delineating the composition and biogenesis of BFP has elucidated the occurrence of two BfpA variants, termed α or β.Citation39 The minor subunits BfpJ, BfpI, and BfpK are also found incorporated in the external BFP filament.Citation40 A vaccine including both BfpA versions and Bfp minor units will likely confer a wider range of protection. The capacity of an EPEC vaccine against BFP subunits to elicit a protective and memorable immune response is untested at this time.

Another strategy targeting adhesins is the rational design of host receptor mimicking saccharides, which is heavily sought after for uropathogenic E. coli (UPEC) infections. The vital role of PapG and FimH in mediating adherence to the uroepithelium makes them prime candidates for antivirulence targeting. Analysis of the binding requirements between PapG and the galabiose receptor guided the design of a PapG inhibitor with a superior aptitude for binding to the host receptor than the natural galabiose receptor; the ability of this PapG inhibitor or derivatives to outcompete the galabiose receptor and consequently disrupt UPEC adherence in vivo is a priority for further development.Citation41 The same strategy is even further along in development for targeting FimH of UPEC’s type 1 pili. FimH inhibitors, or mannosides, are analogous to the FimH receptors and sequester available FimH from binding. In murine models, a mannoside was effective in reducing the bladder bacterial burden during acute and chronic cystitis; furthermore, administration of the mannoside made a trimethoprim–sulfamethoxazole-resistant UPEC strain vulnerable to trimethoprim–sulfamethoxazole treatment.Citation42,Citation43 Cocrystallization of FimH with mannosides facilitated the engineering of a new mannoside with improved affinity for FimH and increased availability in urine, which further reduced bladder bacterial burden compared to the predecessor compound.Citation44

Mannosides have recently been used to target the FimH of adherent-invasive E. coli (AIEC) associated with Crohn’s disease (CD) and E. coli isolated from osteoarticular (hip replacement) infections.Citation45 In a CD murine model, transgenic CEABAC10 mice, which express the human CECAM6 mannosylated receptor, infected with AIEC LF82, were treated with monovalent mannosides post-infection.Citation46 Two mannosides designated 1A-HM and 1CD-HM reduced the bacterial fecal load by ~2.5 and 3 logs, respectively; AIEC LF82 associated with the ileal and colonic mucosa dropped below the limit of detection and mirrored the uninfected mice.Citation46 Furthermore, treatment with 1A-HM and 1CD-HM severely diminished colitis and inflammation.Citation46

Disrupting the biogenesis of adherence factors is another promising AVT. Larzabal et alCitation47 synthesized two small peptides corresponding to the C-terminal coiled-coil domain of the EPEC T3SS translocator EspA. These coiled-coil peptides blocked polymerization of EspA disrupting proper T3SS function and formation of attaching and effacing lesion on HEp-2 cells by EPEC and EHEC.Citation47,Citation48 C57Bl/6 mice inoculated with C. rodentium pretreated with both coiled-coil peptides suffered no damage to their colonic epithelium compared to their untreated counterparts.Citation48

In UPEC, nonpeptidic, pyridone molecules have been designed to bind specifically to the periplasmic chaperone protein, which escorts CUP pili subunits across the periplasmic space.Citation49 The application of chemical synthesis platforms enabled the generation of large-scale 2-pyridone libraries and analysis of various chemical substitutions impacting pilicide potency.Citation50,Citation51 Recently, one pilicide, ec240, was shown to act on the transcriptional level repressing transcription of the fim operon.Citation52 Ec240 is still in the early stages of preclinical development and its direct target(s) remain unknown.

Evasion of the innate immune system and invading the host

Initially, pathogens confront innate immune system barriers, including lysozyme and immunoglobulin A (IgA), and antimicrobial peptides (AMPs).Citation53Citation57 Bacteria, such as Yersinia pestis (the causative agent of pneumonic and bubonic plague) and Moraxella catarrhalis (one of the major causes of otitis media), encode lysozyme inhibitors to combat damage by lysozyme.Citation58,Citation59 Y. pestis mutants with deletion of the ivy gene, which encodes for a lysozyme inhibitor, were attenuated in Y. pestis bubonic and primary pneumonic plague rodent models.Citation59 Wild-type and Δivy Y. pestis strains exhibited indistinguishable virulence potential in FVB lysozyme M-deficient mice, which cannot produce antimicrobial lysozyme LysM.Citation59 In the mucosa, IgA helps maintain balance between the host and their microbiota, as well as, dissuade potential pathogens from invading the mucosa.Citation54 Streptococcus pneumoniae IgA1 protease cleaves IgA1 heavy chain and prevents IgA-dependent killing by human neutrophils in vitro.Citation60 In agreement with this in vitro IgA1 protease activity, mice passively immunized with a human monoclonal IgA1 antibody had enhanced protection from intranasal challenge with an IgA1 protease mutant compared to mice challenged with wild-type S. pneumoniae.Citation60 To counter AMP assault, some pathogens express proteins that proteolytically cleave the AMPs; examples include the EHEC OmpT, C. rodentium CroP, and Y. pestis Pla, which are homologues outer membrane proteases of the omptin family.Citation61Citation64 On the other hand, Haemophilus influenzae imports AMPs from the periplasm, using the ABC transporter SapA, and degrades them for use as nutrient resource.Citation65

In addition to defense mechanisms mounted against innate immune barriers, pathogens can actively manipulate the host immune response. Salmonella enterica ser. Typhimurium is equipped with two T3SSs encoded on separate Salmonella pathogenicity islands (SPI-1 and SPI-2).Citation66Citation68 The SPI-1-encoded T3SS is required for invasion of intestinal epithelial cells, and the SPI-2 system to manipulate phagolysosome fate and promote Salmonella’s intracellular survival.Citation68Citation70 S. enterica secretes TlpA and SseL effector proteins that restrict a nuclear factor κB-mediated inflammatory response and promote bacterial intracellular survival.Citation71,Citation72 Yersinia pseudotuberculosis has a similar anti-inflammatory virulence strategy, the T3SS effector YopJ deubiquitinates IκBα, thereby preventing formation of free nuclear factor κB that would induce a proinflammatory response.Citation73

Supporting the immune system and impeding bacterial invasion

An immune boosting therapeutic approach that is regaining popularity is the administration of exogenous AMPs or compounds that bolster the production of endogenous AMPs. AMP drugs can potentially be used in combination with compounds that derail AMP resistance mechanisms that are common among pathogens.Citation74 Y. pestis Pla and Shigella flexneri IcsP omptins are required for dissemination within the host.Citation75Citation79 The small peptide inhibitor aprotinin is the first inhibitor identified to be capable of inhibiting multiple omptin proteases and provides the basis for the construction of future therapeutic agents that can disrupt the role of omptin proteases in AMP resistance of multiple pathogens.Citation80 More in-depth research is required to understand the numerous AMP resistance mechanisms before an AVT-supporting AMP is successfully composed.

Salicylidene acylhydrazides inhibit the translocation of effector proteins through the T3SSs of both S. enterica ser. Typhimurium and Y. pseudotuberculosis in vitro.Citation81,Citation82 Importantly, salicylidene acylhadrazides also inhibited invasion of epithelial cells by S. enterica ser. Typhimurium in vitro.Citation82,Citation83 These compounds are also effective against Chlamydia trachomatis and S. flexneri T3SSs.Citation84,Citation85 However, the mechanism by which these compounds disrupt different T3SSs remains to be fully elucidated.

Nutrient scavenging

Nutrient acquisition is exceedingly difficult when pathogens have to compete with their fellow microbes and their host begins an attempt to starve them out. Bacterial niche competition can even occur within the same species; E. coli strains MG1655, HS, Nissle 1917, and EDL933 compete with one another for various resources during colonization of the murine intestinal lumen.Citation86Citation88 In the intestinal lumen of specific pathogen-free C57Bl/6 mice, commensal E. coli outcompete C. rodentium.Citation89 Although prevented from colonizing the lumen, C. rodentium invades the intestinal mucosa and infects the epithelial cells inaccessible to commensals.Citation89

The host’s systems are highly reluctant to give up their needed nutrients, such as Fe, and keep them well guarded.Citation90 Host immune cells secrete lipocalin 2 that binds to the E. coli siderophore enterochelin disrupting E. coli’s acquisition of Fe.Citation91 In a Staphylococcus aureus murine infection model, neutrophil calprotectin restricts bacterial growth within abscess by limiting the level of available Mn2+ and Zn2+.Citation90 To survive in their ornery host, pathogens have developed intensive means for gathering their provisions. Pathogens are adept at scavenging valuable essential metals such as Fe, Zn, and Mn. Accumulation of Fe can be toxic to bacteria and requires strict regulation. When intracellular Fe is abundant, the ferric uptake regulator (Fur) binds Fe and represses the transcription of Fe acquisition systems, thereby avoiding Fe overload and toxicity.Citation92 As the Fe level drops, iron-free Fur poorly binds DNA and the once repressed systems become derepressed and more easily transcribed.Citation92 Fur also represses the small noncoding RNA (sRNA) RyhB, which negatively regulates the expression of genes whose products utilize or store Fe.Citation93 S. enterica ser. Typhi fur mutants were defective in their invasion of epithelial and macrophage cells; additionally, S. enterica ser. Typhi requires Fur for in vitro intracellular macrophage survival.Citation94 UPEC CFT073 ΔryhB and ΔryhB Δfur mutants are attenuated in murine bladder infection, while Δfur is not.Citation95 In the absence of RyhB, the corresponding UPEC CFT073 rhyB mutant secreted lower levels of siderophores.Citation95 S. aureus is equipped with iron-regulated surface determinants that allow for the harvesting of Fe from the host’s hemoglobin.Citation96,Citation97 S. aureus scavenges hemoglobin, extracts, and transfers heme to the cytoplasmic IsdG, a heme monooxygenase, which liberates Fe from heme for use by the bacteria.Citation96,Citation98 If too much heme is imported or synthesized by S. aureus, the heme sensory two-component system (HssRS) is activated and mediates the upregulation of the heme response transporter (HtrAB), an efflux pump, to avoid heme toxicity.Citation99Citation101

Starving the enemy

A particularly interesting therapeutic approach to infections is to reestablish healthy microbiota to compete with the disruptive pathogen. After a patient’s gut microbiome is disrupted, Clostridium difficile no longer faces resource competition and takes the opportunity to take over the intestine.Citation102 The modern fecal microbiota transplant (FMT), initially investigated in the treatment of pseudomembranous enterocolitis in 1958, involves the retrieval of the microbiota from a donor’s fecal sample and placed into the intestinal tract of a recipient patient.Citation103 FMT has become popular in recent years for treating C. difficile recurrent infections.Citation104 FMT allows restoration of the microbiota and microbe–microbe competitive interactions. Simplifying the transplantation process and development toward donor pools is likely to contribute to the expansion of FMT use in coming years.Citation105,Citation106 The capacity of FMT to treat various intestinal diseases is yet to be fully explored.

Another “starvation” strategy is the use of small molecules that can potentially be used to target iron uptake systems. Outer membrane transporters rely on energy from the ExbB-ExbD-TonB system to bring in siderophores.Citation107 High-throughput screening of a small molecule library revealed 16 compounds that were bacteriostatic against UPEC under iron-limiting growth conditions.Citation108 A tonB mutant was not sensitive to two of these compounds, suggesting that they disrupted TonB activity.Citation108 These small molecules are still in the early stages of investigation.Citation108 Hma, IreA, IutA, and FyuA are siderophore receptors highly conserved across UPEC strains found in the bacterial outer membrane and surface exposed, which makes them ideal vaccine candidates.Citation109,Citation110 Intranasal immunization with Hma, IreA, IutA, or FyuA elicits a humoral antigen-specific response, and mice were protected against UPEC 1 week after immunization.Citation109,Citation110 A long-lasting response to Hma, IreA, and IutA remains undetermined at this point. FyuA immunization generated long-lived plasma cells indicating a potential for lasting memory and specifically protective against UPEC in pyelonephritis infection models.Citation109,Citation111

Chemical probes are helpful tools for the molecular dissection of biological systems.Citation112 During their investigation of S. aureus HssRS activation by heme, Mike et alCitation113 identified compound ‘882 that indirectly activated HssRS by promoting heme biosynthesis. Independent from HssRS activation, ‘882 was bacteriostatic under fermentative conditions and an ‘882 derivative, ‘373, significantly reduced the bacteria load within liver abscess of S. aureus-infected mice.Citation113 Chemical modification of the parental ‘822 allowed for two different classes of derivatives with activity specific for HssRS activation or inhibitors of fermentative growth.Citation114 While analysis of heme biosynthesis will progress with the usage of HssRS activators, the other new class of compounds represents a promising new scaffold for the development of therapeutics against S. aureus infections.

Microbial population dynamics

A pathogen’s virulence is not solely reliant upon the genetic toolbox equipped within an individual cell. In certain cases, microorganisms will work together in a socialistic fashion to benefit the population at large. This survival strategy includes the formation of biofilms, intracellular bacterial communities (IBCs), and “division of labor” among subpopulations. Biofilms are an accumulation of sessile and diverse microbial populations held together by an extracellular matrix, which shields the microorganisms from antibiotic treatment.Citation115,Citation116 The extracellular matrix consists of an arrangement of various secreted biomolecules potentially including adhesive fibers, carbohydrate-binding proteins, polysaccharides, and extracellular DNA.Citation115,Citation116 Candida albicans and UPEC form biofilms on indwelling medical devices and pose serious health risks.Citation116,Citation117 During infection, UPEC enters superficial umbrella epithelial cells. Intracellular UPEC develops biofilm-like pods and replicates prior to release into the lumen to avoid the exfoliation process of the urothelium.Citation28,Citation118,Citation119 Upon exodus from the surface epithelial cell, UPEC infects additional surface cells, as well as, invading the exposed underlying epithelial cells.Citation28,Citation118,Citation119 Bacteria invading the intermediate epithelium settle quiescent intermediate reservoirs (QIRs) and are safe from antibiotics that abolish the exposed extracelluar.Citation120,Citation121 Weeks later, QIR bacteria can emerge from their fallout shelters and reinitiate the infectious cycle.Citation121,Citation122 IBC formation is not exclusively an UPEC-specific strategy; Klebsiella pneumoniae and Pseudomonas aeruginosa also invade epithelial cells and form intracellular biofilm-like communities.Citation123,Citation124

Within bacterial populations, organized subpopulations are phenotypically distinct and can carry out actions that benefit the entire bacterial community. Y. pseudotuberculosis cells on the peripheral region of a microcolony produce Hmp, a nitric oxide detoxifying enzyme, in response to toxic nitric oxide from the host.Citation125 The Hmp producing subpopulation prevents nitric oxide from penetrating the microcolony, and the protected centroid cells are spared the unnecessary hmp expression.Citation125 Originally recognized for their persistent survival in the presence of penicillin in the early 1940s, persister cells are genotypically identical to their kin yet metabolically dormant.Citation126 The minute persister subpopulation serves as a fail-safe to ensure survival in case of a catastrophic event, such as antibiotic clearance of the metabolically active cells.

Rising evidence indicates that bacterial signaling impacts subpopulation heterogeneity. E. coli cells sensing the presence of indole, which is produced under nutrient-limiting growth conditions, are more likely to enter a persister cell state.Citation127 P. aeruginosa uses quorum sensing (QS) to coordinate the persister subpopulation numbers during logarithmic phase.Citation128 Addition of cell-free spent media from stationary phase cultures or the addition exogenous QS autoinducers pyocyanine or N-(3-oxododecanoyl)-l-homoserine lactone (3OC-HSL) substantially promoted persister cell formation of logarithmic P. aeruginosa cultures.Citation128 The autoinducer competence-stimulating peptide controls the transformational competency and biofilm formation of S. pneumoniae at low concentrations; however, at high concentrations competence-stimulating peptide stalls population growth through stimulating the ComD sensor.Citation129Citation131 These studies highlight the dynamic role of QS in coordinating bacterial populations.

Disturbing the peace of biofilm populations

Biofilm formation allows bacteria to survive antibiotic treatment. Multiple factors are proposed to contribute to biofilm antibiotic tolerance, including persister cell populations, slow growth, and poor antibiotic penetration. Within the past few years, development of platforms for high-throughput screening of chemical libraries has accelerated the discovery of compounds active against C. albicans biofilms.Citation132,Citation133 A family of diazaspirodecane-based compounds was found particularly effective at inhibiting C. albicans biofilm formation.Citation134 Not only did the lead candidate, identified as compound 61894700, disrupt biofilm formation but also filamentation without affecting growth of planktonic cells.Citation134 Prophylactic treatment with compound 61894700 in an oral and invasive systemic candidiasis murine models reduced fungal filamentation and decreased disease severity.Citation134 Another strategy is to find compounds that sensitize biofilms to currently available drugs. Separately, 2-adamantanamine (AC-17) and azoles are not fungistatic, yet the combination of the two is fungicidal.Citation135 In vitro analysis reveals that AC-17-exposed C. albicans had disrupted ergosterol and hyphae production.Citation135 In a guinea pig cutaneous candidiasis model, treatment of subinhibitory fluconazole or AC-17 did not reduce the Candida tissue burden; nevertheless, dual treatment significantly reduced fungal burden.Citation135

UPEC QIR poses the additional challenge of passing the antimicrobial agents through the eukaryotic cells. Accelerating the uroepithelial cell turnover can circumvent this problem by exposing the intracellularly sheltered bacteria. Blango and colleaguesCitation136 found that in a murine model resurgence of UPEC QIRs can be initiated by inducing exfoliation of bladder epithelial cells with chitosan. UPEC UTI89-infected mice that were treated with chitosan had a significant reduction in bacteria loads within the bladder 3 days posttreatment, compared to mice treated with antibiotics alone.Citation136 Chitosan treatment is likely be found to lower the frequency of UPEC UTI recurrence.Citation136 The occurrence of UPEC QIR is reported in multiple clinical studies indicating that these intracellular biofilm-like communities are not specific to murine models.Citation137Citation139 Furthermore, the presence of QIR in human UPEC UTIs validates AVTs, targeting this critical step in pathogenesis.

Biofilm tolerance is largely attributed to the presence of a dormant persister subpopulation.Citation140,Citation141 The persister subpopulation is associated with the recurrent P. aeruginosa infections of cystic fibrosis patients and long-term carriage of Candida by cancer patients prophylactically treated with topical chlorhexidine as part of their oral care.Citation142,Citation143 Therapeutics aimed at “resuscitating” persister cells are of particular importance for preventing recurrent infections. The protease ClpP is normally tightly controlled through ATPases; however, in the presence of acyldepsipeptides, ClpP becomes active independent of ATPases.Citation144 Without proper regulatory control, ClpP nonspecifically degrades bacterial proteins resulting in autodestruction of the bacterial cell.Citation144 In lethal systemic murine infections of E. faecalis and S. aureus, the acyldepsipeptide (ADEP4) rescued 100% and 80% of the mice, respectively.Citation144,Citation145 Unlike rifampicin, ADEP4 eliminated ciprofloxacin surviving S. aureus persister cells in vitro, although a small population of S. aureus survived through clpP null mutations. A combination of rifampicin and ADEP4 effectively abolished S. aureus biofilms in vitro and S. aureus mouse thigh infections.Citation145 This is an example of a combinatorial approach to clear an infection through coordinated targeting of different subpopulations.

Instead of pursuing cells within a microbial population directly, the cells within the population can also be targeted indirectly through severing the intercellular communication systems by which the microbes synchronize their pathogenic efforts. P. aeruginosa coordinates biofilm fate and persister cell population through the synthesis and release of the extracellular autoinducer molecule 3OC-HSL.Citation128,Citation146 Production of both 3OC-HSL and N-butyryl homoserine lactone autoinducers is required for successful lung infection of P. aeruginosa in rodent models.Citation147,Citation148 A class of compound derived from marine algae Delisea pulchra called furanones disrupts the 3OC-HSL sensory system of P. aeruginosa and sensitized biofilms to tobramycin killing in vitro.Citation149 Administration of furanones C30 and C56 extended the survival time of mice with lethal pulmonary P. aeruginosa infections.Citation148,Citation149 By interfering with quorum sensory, furanones disrupt the expression of VFs. This disruption of VFs may potentiate the activity of conventional antibiotics that struggle to clear biofilms.

Regulatory systems of pathogens

To prevent excessive metabolic expenditure, microorganisms closely regulate their core metabolic processes, such as with the fur system previously discussed. Bacterial pathogens can monitor and react to their extracellular environments through two-component sensory systems (TCSs) comprised of a sensor kinase and a response regulator (RR). Typically, upon stimulation, the sensor kinase undergoes autophosphorylation and in turn phosphorylates and activates the RR. The QS E. coli (QseBC) TCS deviates from the customary signal transference cascade. Proper expression of the QseB regulon is tightly controlled through QseC.Citation150,Citation151 Deletion of qseC in UPEC UTI89 leads to virulence attenuation in a murine UTI model plus misregulation of curli, pili, flagella, and core metabolic processes, whereas UPEC UTI ΔqseB and ΔqseBC do not display reduction in virulence.Citation150,Citation151 This contradiction to the conventional TCS model is explained by the ability of QseC to dephosphorylate QseB and allow expression of the QseB repressed genes.Citation151 The PhoPQ TCS is critical for the regulation of VF of S. enterica, UPEC, C. rodentium, and P. aeruginosa.Citation64,Citation152Citation154 Stimulation of the sensor PhoQ by low cation concentrations, acidic pH, and AMP induces canonical activation of the regulatory PhoP.Citation155 S. enterica ser. Typhi strains with deletion of phoPQ were constructed for the development of live attenuated oral vaccine.Citation156,Citation157 Although these strains did not elicit a strong immunogenic response, they were profoundly attenuated in virulence.Citation156,Citation157 The reliance of S. enterica on PhoPQ during infection is attributed to the ability of PhoPQ to coordinate restructuring of the outer membrane.Citation155

sRNAs are an additional regulatory layer of pathogens to control their transcripts. E. coli, P. aeruginosa, S. aureus, and Vibrio cholerae can use sRNA to moderate their production of VFs.Citation158Citation161 In E. coli, the master regulator phoPQ mRNA is regulated by MicA and GcvB, which are sRNAs stabilized by host factor Q.Citation162,Citation163 MicA and GcvB both regulate phoP mRNA translation to different extents, which is reflected in their different effects on the PhoP regulon.Citation163 V. cholerae and Vibrio harveyi have complex regulatory network to modulate their QS pathways. At low cell density and autoinducer concentrations, the QS receptors LuxPQ, LuxN, and CqsS transfer phosphate through LuxU to the RR LuxO.Citation164Citation167 Phosphorylated LuxO promotes the transcription of the four quorum regulatory rna loci (qrr1–4).Citation158,Citation168,Citation169 These Qrr sRNAs redundantly repress and activate the translation of the high cell density HapR and low cell density AphA master regulators, respectively.Citation169Citation172 This regulatory system is reversed in high-density populations with high autoinducer production.Citation169,Citation173 The Vibrio QS regulatory network seems to allow prioritization of VF production. The Qrr sRNAs directly repress the large type 6 secretion system (T6SS) cluster, which encodes the secretory machinery, and indirectly represses T6SS effectors that are activated by HapR.Citation171 The response of Qrr sRNAs genes, such as those encoding for T6SS, directly targeted is relatively more rapid compared to those indirectly regulated through HapR.Citation171,Citation172

Communication breakdown

The disruption of genetic regulatory systems within pathogens poses an option to target multiple virulence systems with one blow and drive the microbe’s virulence to ruin. Misregulation of VF genes results in attenuation. AVTs can block signal detection or inhibit signal relay between the sensor and regulator. The compound LED209 prevents the autophosphorylation of the EHEC sensor QseC.Citation174 Although LED209 treatment failed to prevent EHEC from intestinal colonization in rabbits, LED209 treatment pre- and postinoculation of S. enterica ser. Typhimurium and Franceisella tularensis modestly improved the survival rates of mice.Citation174,Citation175 LED209 demonstrates the feasibility of targeting virulence regulatory systems, and future LED209 derivatives may have improved bioavailability and efficacy during infection.Citation174,Citation175 The QseBC system cross-interacts with the PmrAB system associated with polymyxin resistance.Citation176 In the absence of QseC, PmrB favorably phosphorylates QseB, which would normally be dephosphorylated by QseC.Citation176 QseC and PmrB balance out the level of active QseB. This delicate harmonization is reflected during UPEC infection models. In a UPEC model for acute UTI, UPEC UTI89 ΔqseC forms significantly fewer IBCs than wild type, and deletion of pmrB in the UTI89 ΔqseC strain restores IBC abundance to wild-type levels.Citation176,Citation177 An AVT that promotes the buildup of QseB to diminish UPEC virulence by increasing phosphorylated QseB through enhancing the PmrB kinase and/or inhibiting QseC phosphatase activities would be ideal.Citation176

The Vibrio QS network contains multiple promising targets, including signal receptors and gene regulators.Citation178 A pro-QS strategy is proposed to disrupt the QS signaling cascade and expression of VF between low and high cell density population transitions.Citation179,Citation180 One class of compounds was found that inhibits the ATPase action of phosphorylated LuxO and consequently disrupted regulation of the downstream QS circuitry.Citation178 Analysis of the biosynthesis of the autoinducer CAI-1 identified the intermediate product Ea-CAI-1 as more potent agonist than CAI-1.Citation181 Recently, stable derivatives of Ea-CAI-1 were developed. The most stable Ea-CAI-1 derivative, compound 18, is approximately tenfold more potent than CAI-1.Citation179 Compound 18 does not display toxicity to murine fibroblast cells or disrupt the growth of V. cholerae.Citation179 The ability of compound 18 to improve V. cholerae infection remains to be tested. These studies demonstrate the validity of prosignaling therapeutics strategies, promoting the suppression of VFs. These AVTs may also be applicable to other sensory systems, such as with the MerR-like transcriptional regulator BluR in E. coli. BluR is a repressor of ymgA and ymgB.Citation182 In the presence of blue-light irradiation, BluF, a flavin binding sensor, binds and antagonizes BluR allowing for the transcription of ymgA and ymgB, which promote biofilm formation through the RcsCDB phosphorelay system.Citation182 Blue-light irradiation was recently shown to deter growth of E. coli in a strain and growth phase-dependent manner, as well as, induce a persister-like state in some of the cells, which may involve BluFR light sensing (Mitchell et al, unpublished data, 2016). In sum, the targeting of virulence regulation both at the transcriptional and translational levels is a very promising prospect that has many avenues open for exploration and development.

Conclusion and outlook

Commensal microorganisms maintain a delicate balance with their host. Pathogenic microorganisms acquire VF stochastically and via selective pressures and use them to misbehave in a niche-dependent manner. During the 20th century, classical antimicrobials have helped to strike back at pathogens; nevertheless, the pathogen menace is equipped with strategies to resist antimicrobials and return for their revenge. The widespread usage of antimicrobials awakened a force of resistance mechanisms, which we must confront to thwart the postantibiotic era we are facing. The quagmire of antibiotic resistance has reached such an extraordinary level that it has finally captured the interest of politicians. In 2014, US President Obama signed Executive Order 13676: Combating Antibiotic-Resistant Bacteria, which called for the formation of Task Force for Combatting Antimicrobial-Resistant Bacteria.Citation183 The plan of action outlined by the task force set in place under this executive order prioritizes the development of alternative antimicrobials, such as AVTs, and next-generation diagnostic tools.Citation184 We are at a critical point where necessity is motivating the development of new discovery platforms. Many questions remain to be answered. AVTs represent a new hope for starting a silver age of antibiotics.

Disclosure

The authors report no conflicts of interest in this work.

References

  • FlemingAOn the antibacterial action of cultures of a penicillium, with special reference to their use in the isolation of B. influenzaeBr J Exp Pathol1929103226236
  • WainwrightMKristiansenJEOn the 75th anniversary of prontosilDyes Pigm2011883231234
  • DomagkGEin beitrag zur chemotherapie der bakteriellen Infektionen [A contribution to the chemotherapy of bacterial infections]DMW19356107250253 German
  • KampmeierRHIntroduction of sulfonamide therapy for gonorrheaSex Transm Dis198310281846362039
  • DunlopEMGonorrhoea and the sulphonamidesBr J Vener Dis1949252818318151908
  • ChainEFloreyHWGardnerADHeatleyNGPenicillin as a chemotherapeutic agentLancet19402366104226228
  • WillcoxRRA survey of problems in the antibiotic treatment of gonorrhoea. With special reference to South-East AsiaBr J Vener Dis19704632172424246923
  • AshfordWAGolashRGHemmingVGPenicillinase-producing Neisseria gonorrhoeaeLancet19762798765765860519
  • PhillipsIB-Lactamase-producing, penicillin-resistant gonococcusLancet1976308798765665760518
  • WorkowskiKABermanSMDouglasJMJrEmerging antimicrobial resistance in Neisseria gonorrhoeae: urgent need to strengthen prevention strategiesAnn Intern Med2008148860661318413622
  • WorkowskiKABolanGACenters for Disease Control and PreventionSexually transmitted diseases treatment guidelines, 2015MMWR Recomm Rep201564RR–03113726042815
  • LingLLSchneiderTPeoplesAJA new antibiotic kills pathogens without detectable resistanceNature2015517753545545925561178
  • RaskoDASperandioVAnti-virulence strategies to combat bacteria-mediated diseaseNat Rev Drug Discov20109211712820081869
  • ClearyJLaiLCShawRKEnteropathogenic Escherichia coli (EPEC) adhesion to intestinal epithelial cells: role of bundle-forming pili (BFP), EspA filaments and intiminMicrobiology2004150pt 352753814993302
  • DaniellSJTakahashiNWilsonRThe filamentous type III secretion translocon of enteropathogenic Escherichia coliCell Microbiol200131286587111736997
  • GironJAHoASSchoolnikGKAn inducible bundle-forming pilus of enteropathogenic Escherichia coliScience199125450327107131683004
  • BieberDRamerSWWuCYType IV pili, transient bacterial aggregates, and virulence of enteropathogenic Escherichia coliScience19982805372211421189641917
  • DengWVallanceBALiYPuenteJLFinlayBBCitrobacter rodentium translocated intimin receptor (Tir) is an essential virulence factor needed for actin condensation, intestinal colonization and colonic hyperplasia in miceMol Microbiol20034819511512657048
  • DeVinneyRSteinMReinscheidDAbeARuschkowskiSFinlayBBEnterohemorrhagic Escherichia coli O157: H7 produces Tir, which is translocated to the host cell membrane but is not tyrosine phosphorylatedInfect Immun19996752389239810225900
  • KennyBDeVinneyRSteinMReinscheidDJFreyEAFinlayBBEnteropathogenic E. coli (EPEC) transfers its receptor for intimate adherence into mammalian cellsCell19979145115209390560
  • MiyazakiJBa-TheinWKumaoTAkazaHHayashiHIdentification of a type III secretion system in uropathogenic Escherichia coliFEMS Microbiol Lett2002212222122812113938
  • SubashchandraboseSHazenTHRaskoDAMobleyHLDraft genome sequences of five recent human uropathogenic Escherichia coli isolatesPathog Dis20136916670
  • MartinezJJMulveyMASchillingJDPinknerJSHultgrenSJType 1 pilus-mediated bacterial invasion of bladder epithelial cellsEMBO J200019122803281210856226
  • MellataMDho-MoulinMDozoisCMRole of virulence factors in resistance of avian pathogenic Escherichia coli to serum and in pathogenicityInfect Immun200371153654012496207
  • StacyAKMitchellNMMadduxJTEvaluation of the prevalence and production of Escherichia coli common pilus among avian pathogenic E. coli and its role in virulencePLoS One201491e8656524466152
  • KorhonenTKValtonenMVParkkinenJSerotypes, hemolysin production, and receptor recognition of Escherichia coli strains associated with neonatal sepsis and meningitisInfect Immun19854824864912580792
  • WaksmanGHultgrenSJStructural biology of the chaperone-usher pathway of pilus biogenesisNat Rev Microbiol200971176577419820722
  • MulveyMALopez-BoadoYSWilsonCLInduction and evasion of host defenses by type 1-piliated uropathogenic Escherichia coliScience19982825393149414979822381
  • KalleniusGMollbyRSvensonSBOccurrence of P-fimbriated Escherichia coli in urinary tract infectionsLancet198128260–8261136913726171697
  • NorgrenMNormarkSLarkDMutations in E. coli cistrons affecting adhesion to human cells do not abolish Pap pili fiber formationEMBO J198435115911656145589
  • NormarkSLarkDHullRGenetics of digalactoside-binding adhesin from a uropathogenic Escherichia coli strainInfect Immun19834139429496136465
  • HultgrenSJLindbergFMagnussonGKihlbergJTennentJMNormarkSThe PapG adhesin of uropathogenic Escherichia coli contains separate regions for receptor binding and for the incorporation into the pilusProc Natl Acad Sci U S A19898612435743612567514
  • LefflerHEdénCSChemical identification of a glycosphingolipid receptor for Escherichia coli attaching to human urinary tract epithelial cells and agglutinating human erythrocytesFEMS Microbiol Lett198083127134
  • VaisanenVEloJTallgrenLGMannose-resistant haemagglutination and P antigen recognition are characteristic of Escherichia coli causing primary pyelonephritisLancet198128260–8261136613696171696
  • NilssonUStrikerRTHultgrenSJMagnussonGPapG adhesin from E. coli J96 recognizes the same saccharide epitope when present on whole bacteria and as isolated proteinBioorg Med Chem1996411180918179007266
  • SchrieferAMaltezJRSilvaNStoeckleMYBarral-NettoMRileyLWExpression of a pilin subunit BfpA of the bundle-forming pilus of enteropathogenic Escherichia coli in an aroA live Salmonella vaccine strainVaccine1999177–877077810067682
  • LevineMMEscherichia coli that cause diarrhea: enterotoxigenic, enteropathogenic, enteroinvasive, enterohemorrhagic, and enteroadherentJ Infect Dis198715533773893543152
  • MartinezMBTaddeiCRRuiz-TagleATrabulsiLRGironJAAntibody response of children with enteropathogenic Escherichia coli infection to the bundle-forming pilus and locus of enterocyte effacement-encoded virulence determinantsJ Infect Dis199917912692749841853
  • BlankTEZhongHBellALWhittamTSDonnenbergMSMolecular variation among type IV Pilin (bfpA) genes from diverse enteropathogenic Escherichia coli strainsInfect Immun200068127028703811083828
  • Martinez de la PenaCFDe MasiLNisaSBfpI, BfpJ, and BfpK minor pilins are important for the function and biogenesis of bundle-forming pili expressed by enteropathogenic Escherichia coliJ Bacteriol2015198584685626712935
  • OhlssonJJassJUhlinBEKihlbergJNilssonUJDiscovery of potent inhibitors of PapG adhesins from uropathogenic Escherichia coli through synthesis and evaluation of galabiose derivativesChembiochem20023877277912203976
  • CusumanoCKPinknerJSHanZTreatment and prevention of urinary tract infection with orally active FimH inhibitorsSci Transl Med20113109109ra115
  • TotsikaMKostakiotiMHannanTJA FimH inhibitor prevents acute bladder infection and treats chronic cystitis caused by multidrug-resistant uropathogenic Escherichia coli ST131J Infect Dis2013208692192823737602
  • JarvisCHanZKalasVAntivirulence isoquinolone mannosides: optimization of the biaryl aglycone for FimH lectin binding affinity and efficacy in the treatment of chronic UTIChemMedChem201611436737326812660
  • ChalopinTBrissonnetYSivignonAInhibition profiles of mono-and polyvalent FimH antagonists against 10 different Escherichia coli strainsOrg Biomol Chem20151346113691137526440382
  • SivignonAYanXAlvarez DortaDDevelopment of heptylmannoside-based glycoconjugate antiadhesive compounds against adherent-invasive Escherichia coli bacteria associated with Crohn’s diseaseMBio201566e1298e1315
  • LarzabalMMercadoECVilteDASalazar-GonzalezHGataldiANavarro-GarciaFDesigned coiled-coil peptides inhibit the type three secretion system of enteropathogenic Escherichia coliPLoS One201052e904620140230
  • LarzábalMZottaEIbarraCEffect of coiled-coil peptides on the function of the type III secretion system-dependent activity of enterohemorragic Escherichia coli O157: H7 and Citrobacter rodentiumInt J Med Microbiol2013303191523312797
  • SvenssonALarssonAEmtenasHDesign and evaluation of pilicides: potential novel antibacterial agents directed against uropathogenic Escherichia coliChembiochem200121291591811948880
  • HanZPinknerJSFordBLead optimization studies on FimH antagonists: discovery of potent and orally bioavailable ortho-substituted biphenyl mannosidesJ Med Chem20125583945395922449031
  • ChorellEPinknerJSPhanGDesign and synthesis of C-2 substituted thiazolo and dihydrothiazolo ring-fused 2-pyridones: pilicides with increased antivirulence activityJ Med Chem201053155690569520586493
  • GreeneSEPinknerJSChorellEPilicide ec240 disrupts virulence circuits in uropathogenic Escherichia coliMBio201456e0203825352623
  • FlemingAAllisonVDObservations on a bacteriolytic substance (“lysozyme”) found in secretions and tissuesBr J Exp Pathol192235252260
  • MacphersonAJMcCoyKDJohansenFEBrandtzaegPThe immune geography of IgA induction and functionMucosal Immunol200811112219079156
  • RussellJPDiamondGTarverAPScanlinTFBevinsCLCoordinate induction of two antibiotic genes in tracheal epithelial cells exposed to the inflammatory mediators lipopolysaccharide and tumor necrosis factor alphaInfect Immun1996645156515688613361
  • VoraPYoudimAThomasLSβ-defensin-2 expression is regulated by TLR signaling in intestinal epithelial cellsJ Immunol200417395398540515494486
  • Rivas-SantiagoBHernandez-PandoRCarranzaCExpression of cathelicidin LL-37 during Mycobacterium tuberculosis infection in human alveolar macrophages, monocytes, neutrophils, and epithelial cellsInfect Immun200876393594118160480
  • JoslinSNPybusCLabandeira-ReyMA Moraxella catarrhalis two-component signal transduction system necessary for growth in liquid media affects production of two lysozyme inhibitorsInfect Immun201583114616025312959
  • DerbiseAPierreFMerchezMInheritance of the lysozyme inhibitor Ivy was an important evolutionary step by Yersinia pestis to avoid the host innate immune responseJ Infect Dis2013207101535154323402825
  • JanoffENRubinsJBFaschingCPneumococcal IgA1 protease subverts specific protection by human IgA1Mucosal Immunol20147224925623820749
  • GalvanEMLasaroMASchifferliDMCapsular antigen fraction 1 and Pla modulate the susceptibility of Yersinia pestis to pulmonary antimicrobial peptides such as cathelicidinInfect Immun20087641456146418227173
  • ThomassinJLBrannonJRGibbsBFGruenheidSLe MoualHOmpT outer membrane proteases of enterohemorrhagic and enteropathogenic Escherichia coli contribute differently to the degradation of human LL-37Infect Immun201280248349222144482
  • BrannonJRThomassinJLGruenheidSLe MoualHAntimicrobial peptide conformation as a structural determinant of omptin protease specificityJ Bacteriol2015197223583359126350132
  • Le SageVZhuLLepageCAn outer membrane protease of the omptin family prevents activation of the Citrobacter rodentium PhoPQ two-component system by antimicrobial peptidesMol Microbiol20097419811119708916
  • SheltonCLRaffelFKBeattyWLJohnsonSMMasonKMSap transporter mediated import and subsequent degradation of antimicrobial peptides in HaemophilusPLoS Pathog2011711e100236022072973
  • HenselMSheaJERaupachBFunctional analysis of ssaJ and the ssaK/U operon, 13 genes encoding components of the type III secretion apparatus of Salmonella pathogenicity island 2Mol Microbiol19972411551679140973
  • OchmanHSonciniFCSolomonFGroismanEAIdentification of a pathogenicity island required for Salmonella survival in host cellsProc Natl Acad Sci U S A19969315780078048755556
  • GalanJECurtissRCloning and molecular characterization of genes whose products allow Salmonella typhimurium to penetrate tissue culture cellsProc Natl Acad Sci U S A19898616638363872548211
  • CirilloDMValdiviaRHMonackDMFalkowSMacrophage-dependent induction of the Salmonella pathogenicity island 2 type III secretion system and its role in intracellular survivalMol Microbiol19983011751889786194
  • PfeiferCGMarcusSLSteele-MortimerOKnodlerLAFinlayBBSalmonella typhimurium virulence genes are induced upon bacterial invasion into phagocytic and nonphagocytic cellsInfect Immun199967115690569810531217
  • NewmanRMSalunkhePGodzikAReedJCIdentification and characterization of a novel bacterial virulence factor that shares homology with mammalian Toll/interleukin-1 receptor family proteinsInfect Immun200674159460116369016
  • MukherjeeSOrthKKrajewskiSSalmonella secreted factor L deubiquitinase of Salmonella typhimurium inhibits NF-κB, suppresses IκBα ubiquitination and modulates innate immune responsesJ Immunol200818075045505618354230
  • ZhouHMonackDMKayagakiNWertzIYersinia virulence factor YopJ acts as a deubiquitinase to inhibit NF-κB activationJ Exp Med2005202101327133216301742
  • Le MoualHThomassinJ-LBrannonJRAntimicrobial peptides as an alternative approach to treat bacterial infectionsJ Clin Cell Immunol2013S13004
  • ShereKDSallustioSManessisAD’AversaTGGoldbergMBDisruption of IcsP, the major Shigella protease that cleaves IcsA, accelerates actin-based motilityMol Microbiol19972534514629302008
  • WelkosSLFriedlanderAMDavisKJStudies on the role of plasminogen activator in systemic infection by virulent Yersinia pestis strain C092Microb Pathog19972342112239344782
  • SodeindeOASubrahmanyamYVStarkKQuanTBaoYGoguenJDA surface protease and the invasive character of plagueScience19922585084100410071439793
  • EgileCd’HautevilleHParsotCSansonettiPJSopA, the outer membrane protease responsible for polar localization of IcsA in Shigella flexneriMol Microbiol1997235106310739076742
  • LathemWWPricePAMillerVLGoldmanWEA plasminogen-activating protease specifically controls the development of primary pneumonic plagueScience2007315581150951317255510
  • BrannonJRBurkDLLeclercJMInhibition of outer membrane proteases of the omptin family by aprotininInfect Immun20158362300231125824836
  • NordfelthRKauppiAMNorbergHAWolf-WatzHElofssonMSmall-molecule inhibitors specifically targeting type III secretionInfect Immun20057353104311415845518
  • HudsonDLLaytonANFieldTRInhibition of type III secretion in Salmonella enterica serovar Typhimurium by small-molecule inhibitorsAntimicrob Agents Chemother20075172631263517502403
  • NegreaABjurEYgbergSEElofssonMWolf-WatzHRhenMSalicylidene acylhydrazides that affect type III protein secretion in Salmonella enterica serovar typhimuriumAntimicrob Agents Chemother20075182867287617548496
  • MuschiolSBaileyLGylfeAA small-molecule inhibitor of type III secretion inhibits different stages of the infectious cycle of Chlamydia trachomatisProc Natl Acad Sci U S A200610339145661457116973741
  • VeenendaalAKSundinCBlockerAJSmall-molecule type III secretion system inhibitors block assembly of the Shigella type III secretonJ Bacteriol2009191256357018996990
  • MomoseYHirayamaKItohKCompetition for proline between indigenous Escherichia coli and E. coli O157:H7 in gnotobiotic mice associated with infant intestinal microbiota and its contribution to the colonization resistance against E. coli O157:H7Antonie Van Leeuwen-hoek2008942165171
  • MomoseYHirayamaKItohKEffect of organic acids on inhibition of Escherichia coli O157:H7 colonization in gnotobiotic mice associated with infant intestinal microbiotaAntonie Van Leeuwenhoek2008931–214114917674138
  • FabichAJJonesSAChowdhuryFZComparison of carbon nutrition for pathogenic and commensal Escherichia coli strains in the mouse intestineInfect Immun20087631143115218180286
  • KamadaNKimYGShamHPRegulated virulence controls the ability of a pathogen to compete with the gut microbiotaScience201233660861325132922582016
  • CorbinBDSeeleyEHRaabAMetal chelation and inhibition of bacterial growth in tissue abscessesScience2008319586596296518276893
  • FloTHSmithKDSatoSLipocalin 2 mediates an innate immune response to bacterial infection by sequestrating ironNature2004432701991792115531878
  • LeeJWHelmannJDFunctional specialization within the Fur family of metalloregulatorsBiometals2007203–448549917216355
  • MasseEGottesmanSA small RNA regulates the expression of genes involved in iron metabolism in Escherichia coliProc Natl Acad Sci U S A20029974620462511917098
  • LeclercJMDozoisCMDaigleFRole of the Salmonella enterica serovar Typhi Fur regulator and small RNAs RfrA and RfrB in iron homeostasis and interaction with host cellsMicrobiology2013159pt 359160223306672
  • PorcheronGHabibRHouleSThe small RNA RyhB contributes to siderophore production and virulence of uropathogenic Escherichia coliInfect Immun201482125056506825245805
  • MazmanianSKSkaarEPGasparAHPassage of heme-iron across the envelope of Staphylococcus aureusScience2003299560890690912574635
  • TorresVJPishchanyGHumayunMSchneewindOSkaarEPStaphylococcus aureus IsdB is a hemoglobin receptor required for heme iron utilizationJ Bacteriol2006188248421842917041042
  • SkaarEPGasparAHSchneewindOIsdG and IsdI, heme-degrading enzymes in the cytoplasm of Staphylococcus aureusJ Biol Chem2004279143644314570922
  • FriedmanDBStauffDLPishchanyGWhitwellCWTorresVJSkaarEPStaphylococcus aureus redirects central metabolism to increase iron availabilityPLoS Pathog200628e8716933993
  • StauffDLTorresVJSkaarEPSignaling and DNA-binding activities of the Staphylococcus aureus HssR-HssS two-component system required for heme sensingJ Biol Chem200728236261112612117635909
  • TorresVJStauffDLPishchanyGA Staphylococcus aureus regulatory system that responds to host heme and modulates virulenceCell Host Microbe20071210911918005689
  • GhoseCClostridium difficile infection in the twenty-first centuryEmerg Microbes Infect201329e6226038491
  • EisemanBSilenWBascomGSKauvarAJFecal enema as an adjunct in the treatment of pseudomembranous enterocolitisSurgery195844585485913592638
  • MattilaEUusitalo-SeppalaRWuorelaMFecal transplantation, through colonoscopy, is effective therapy for recurrent Clostridium difficile infectionGastroenterology2012142349049622155369
  • YoungsterIHohmannELFecal microbiota transplantation for Clostridium difficile infection – replyJAMA2015313772625688788
  • HamiltonMJWeingardenARSadowskyMJKhorutsAStandardized frozen preparation for transplantation of fecal microbiota for recurrent Clostridium difficile infectionAm J Gastroenterol2012107576176722290405
  • NoinajNGuillierMBarnardTJBuchananSKTonB-dependent transporters: regulation, structure, and functionAnnu Rev Microbiol201064436020420522
  • YepAMcQuadeTKirchhoffPLarsenMMobleyHLInhibitors of TonB function identified by a high-throughput screen for inhibitors of iron acquisition in uropathogenic Escherichia coli CFT073MBio201452e1089e1113
  • BrumbaughARSmithSNMobleyHLImmunization with the yersiniabactin receptor, FyuA, protects against pyelonephritis in a murine model of urinary tract infectionInfect Immun20138193309331623798537
  • AlteriCJHaganECSivickKESmithSNMobleyHLMucosal immunization with iron receptor antigens protects against urinary tract infectionPLoS Pathog200959e100058619806177
  • BrumbaughARSmithSNSubashchandraboseSBlocking yersiniabactin import attenuates extraintestinal pathogenic Escherichia coli in cystitis and pyelonephritis and represents a novel target to prevent urinary tract infectionInfect Immun20158341443145025624354
  • ArrowsmithCHAudiaJEAustinCThe promise and peril of chemical probesNat Chem Biol201511853654126196764
  • MikeLADutterBFStauffDLActivation of heme biosynthesis by a small molecule that is toxic to fermenting Staphylococcus aureusProc Natl Acad Sci U S A2013110208206821123630262
  • DutterBFMikeLAReidPRDecoupling activation of heme biosynthesis from anaerobic toxicity in a molecule active in Staphylococcus aureusACS Chem Biol Epub201634
  • KostakiotiMHadjifrangiskouMHultgrenSJBacterial biofilms: development, dispersal, and therapeutic strategies in the dawn of the postantibiotic eraCold Spring Harb Perspect Med201334a01030623545571
  • JacobsenSMSticklerDJMobleyHLShirtliffMEComplicated catheter-associated urinary tract infections due to Escherichia coli and Proteus mirabilisClin Microbiol Rev2008211265918202436
  • KojicEMDarouicheROCandida infections of medical devicesClin Microbiol2004172255267
  • AndersonGGPalermoJJSchillingJDRothRHeuserJHultgrenSJIntracellular bacterial biofilm-like pods in urinary tract infectionsScience2003301562910510712843396
  • JusticeSSHungCTheriotJADifferentiation and developmental pathways of uropathogenic Escherichia coli in urinary tract pathogenesisProc Natl Acad Sci U S A200410151333133814739341
  • BlangoMGMulveyMAPersistence of uropathogenic Escherichia coli in the face of multiple antibioticsAntimicrob Agents Chemother20105451855186320231390
  • SchillingJDLorenzRGHultgrenSJEffect of trimethoprim-sulfamethoxazole on recurrent bacteriuria and bacterial persistence in mice infected with uropathogenic Escherichia coliInfect Immun200270127042704912438384
  • MulveyMASchillingJDHultgrenSJEstablishment of a persistent Escherichia coli reservoir during the acute phase of a bladder infectionInfect Immun20016974572457911402001
  • Garcia-MedinaRDunneWMSinghPKBrodySLPseudomonas aeruginosa acquires biofilm-like properties within airway epithelial cellsInfect Immun200573128298830516299327
  • RosenDAPinknerJSJonesJMWalkerJNCleggSHultgrenSJUtilization of an intracellular bacterial community pathway in Klebsiella pneumoniae urinary tract infection and the effects of FimK on type 1 pilus expressionInfect Immun20087673337334518411285
  • DavisKMMohammadiSIsbergRRCommunity behavior and spatial regulation within a bacterial microcolony in deep tissue sites serves to protect against host attackCell Host Microbe2015171213125500192
  • BiggerJTreatment of staphylococcal infections with penicillin by intermittent sterilisationLancet19442446320497500
  • VegaNMAllisonKRKhalilASCollinsJJSignaling-mediated bacterial persister formationNat Chem Biol20128543143322426114
  • MokerNDeanCRTaoJPseudomonas aeruginosa increases formation of multidrug-tolerant persister cells in response to quorum-sensing signaling moleculesJ Bacteriol201019271946195520097861
  • OggioniMRTrappettiCKadiogluASwitch from planktonic to sessile life: a major event in pneumococcal pathogenesisMol Microbiol20066151196121016925554
  • HavarsteinLSCoomaraswamyGMorrisonDAAn unmodified heptadecapeptide pheromone induces competence for genetic transformation in Streptococcus pneumoniaeProc Natl Acad Sci U S A1995922411140111447479953
  • OggioniMRIannelliFRicciSAntibacterial activity of a competence-stimulating peptide in experimental sepsis caused by Streptococcus pneumoniaeAntimicrob Agents Chemother200448124725473215561850
  • LaFleurMDLucumiENapperADDiamondSLLewisKNovel high-throughput screen against Candida albicans identifies antifungal potentiators and agents effective against biofilmsJ Antimicrob Chemother201166482082621393183
  • FazlyAJainCDehnerACChemical screening identifies filastatin, a small molecule inhibitor of Candida albicans adhesion, morphogenesis, and pathogenesisProc Natl Acad Sci U S A201311033135941359923904484
  • PierceCGChaturvediAKLazzellALA novel small molecule inhibitor of Candida albicans biofilm formation, filamentation and virulence with low potential for the development of resistance. NPJ Biofilms MicrobiomesEpub20158121
  • LafleurMDSunLListerIPotentiation of azole antifungals by 2-adamantanamineAntimicrob Agents Chemother20135783585359223689724
  • BlangoMGOttEMErmanAVeranicPMulveyMAForced resurgence and targeting of intracellular uropathogenic Escherichia coli reservoirsPLoS One201493e9332724667805
  • RobinoLScavonePAraujoLAlgortaGZuninoPVignoliRDetection of intracellular bacterial communities in a child with Escherichia coli recurrent urinary tract infectionsPathog Dis2013683788123733378
  • KhasriyaRSathiananthamoorthySIsmailSSpectrum of bacterial colonization associated with urothelial cells from patients with chronic lower urinary tract symptomsJ Clin Microbiol20135172054206223596238
  • RosenDAHootonTMStammWEHumphreyPAHultgrenSJDetection of intracellular bacterial communities in human urinary tract infectionPLoS Med2007412e32918092884
  • SpoeringALLewisKBiofilms and planktonic cells of Pseudomonas aeruginosa have similar resistance to killing by antimicrobialsJ Bacteriol2001183236746675111698361
  • LaFleurMDKumamotoCALewisKCandida albicans biofilms produce antifungal-tolerant persister cellsAntimicrob Agents Chemother200650113839384616923951
  • LafleurMDQiQLewisKPatients with long-term oral carriage harbor high-persister mutants of Candida albicansAntimicrob Agents Chemother2010541394419841146
  • MulcahyLRBurnsJLLorySLewisKEmergence of Pseudomonas aeruginosa strains producing high levels of persister cells in patients with cystic fibrosisJ Bacteriol2010192236191619920935098
  • Brotz-OesterheltHBeyerDKrollHPDysregulation of bacterial proteolytic machinery by a new class of antibioticsNat Med200511101082108716200071
  • ConlonBPNakayasuESFleckLEActivated ClpP kills persisters and eradicates a chronic biofilm infectionNature2013503747636537024226776
  • DaviesDGParsekMRPearsonJPIglewskiBHCostertonJWGreenbergEPThe involvement of cell-to-cell signals in the development of a bacterial biofilmScience199828053612952989535661
  • PearsonJPFeldmanMIglewskiBHPrinceAPseudomonas aeruginosa cell-to-cell signaling is required for virulence in a model of acute pulmonary infectionInfect Immun20006874331433410858254
  • WuHSongZHentzerMSynthetic furanones inhibit quorum-sensing and enhance bacterial clearance in Pseudomonas aeruginosa lung infection in miceJ Antimicrob Chemother20045361054106115117922
  • HentzerMWuHAndersenJBAttenuation of Pseudomonas aeruginosa virulence by quorum sensing inhibitorsEMBO J200322153803381512881415
  • HadjifrangiskouMKostakiotiMChenSLHendersonJPGreeneSEHultgrenSJA central metabolic circuit controlled by QseC in pathogenic Escherichia coliMol Microbiol20118061516152921542868
  • KostakiotiMHadjifrangiskouMPinknerJSHultgrenSJQseC-mediated dephosphorylation of QseB is required for expression of genes associated with virulence in uropathogenic Escherichia coliMol Microbiol20097361020103119703104
  • AlteriCJLindnerJRReissDJSmithSNMobleyHLThe broadly conserved regulator PhoP links pathogen virulence and membrane potential in Escherichia coliMol Microbiol201182114516321854465
  • MacfarlaneELKwasnickaAHancockRERole of Pseudomonas aeruginosa PhoP-phoQ in resistance to antimicrobial cationic peptides and aminoglycosidesMicrobiology2000146pt 102543255411021929
  • MillerSIKukralAMMekalanosJJA two-component regulatory system (phoP phoQ) controls Salmonella typhimurium virulenceProc Natl Acad Sci U S A19898613505450582544889
  • DalebrouxZDMillerSISalmonellae PhoPQ regulation of the outer membrane to resist innate immunityCurr Opin Microbiol20141710611324531506
  • HohmannELOlettaCAKilleenKPMillerSIphoP/phoQ-deleted Salmonella typhi (Ty800) is a safe and immunogenic single dose typhoid fever vaccine in volunteersJ Infect Dis19961736140814148648213
  • HohmannELOlettaCAMillerSIEvaluation of a phoP/phoQ-deleted, aroA-deleted live oral Salmonella typhi vaccine strain in human volunteersVaccine199614119248821644
  • LenzDHMokKCLilleyBNKulkarniRVWingreenNSBasslerBLThe small RNA chaperone Hfq and multiple small RNAs control quorum sensing in Vibrio harveyi and Vibrio choleraeCell20041181698215242645
  • LenzDHMillerMBZhuJKulkarniRVBasslerBLCsrA and three redundant small RNAs regulate quorum sensing in Vibrio choleraeMol Microbiol20055841186120216262799
  • PichonCFeldenBSmall RNA genes expressed from Staphylococcus aureus genomic and pathogenicity islands with specific expression among pathogenic strainsProc Natl Acad Sci U S A200510240142491425416183745
  • WildermanPJSowaNAFitzGeraldDJIdentification of tandem duplicate regulatory small RNAs in Pseudomonas aeruginosa involved in iron homeostasisProc Natl Acad Sci U S A2004101269792979715210934
  • CoornaertALuAMandinPSpringerMGottesmanSGuillierMMicA sRNA links the PhoP regulon to cell envelope stressMol Microbiol201076246747920345657
  • CoornaertAChiaruttiniCSpringerMGuillierMPost-transcriptional control of the Escherichia coli PhoQ-PhoP two-component system by multiple sRNAs involves a novel pairing region of GcvBPLoS Genet201391e100315623300478
  • FreemanJABasslerBLA genetic analysis of the function of LuxO, a two-component response regulator involved in quorum sensing in Vibrio harveyiMol Microbiol199931266567710027982
  • FreemanJABasslerBLSequence and function of LuxU: a two-component phosphorelay protein that regulates quorum sensing in Vibrio harveyiJ Bacteriol19991838999069922254
  • NeiditchMBFederleMJPompeaniAJLigand-induced asymmetry in histidine sensor kinase complex regulates quorum sensingCell200612661095110816990134
  • SwemLRSwemDLO’LoughlinCTA quorum-sensing antagonist targets both membrane-bound and cytoplasmic receptors and controls bacterial pathogenicityMol Cell200935214315319647512
  • LilleyBNBasslerBLRegulation of quorum sensing in Vibrio harveyi by LuxO and sigma-54Mol Microbiol200036494095410844680
  • TuKCBasslerBLMultiple small RNAs act additively to integrate sensory information and control quorum sensing in Vibrio harveyiGenes Dev200721222123317234887
  • RutherfordSTValastyanJSTaillefumierTWingreenNSBasslerBLComprehensive analysis reveals how single nucleotides contribute to noncoding RNA function in bacterial quorum sensingProc Natl Acad Sci U S A201511244E6038E604726483489
  • ShaoYBasslerBLQuorum regulatory small RNAs repress type VI secretion in Vibrio choleraeMol Microbiol201492592193024698180
  • ShaoYFengLRutherfordSTPapenfortKBasslerBLFunctional determinants of the quorum-sensing non-coding RNAs and their roles in target regulationEMBO J201332152158217123838640
  • RutherfordSTvan KesselJCShaoYBasslerBLAphA and LuxR/HapR reciprocally control quorum sensing in vibriosGenes Dev201125439740821325136
  • RaskoDAMoreiraCGLi deRTargeting QseC signaling and virulence for antibiotic developmentScience200832158921078108018719281
  • CurtisMMRussellRMoreiraCGQseC inhibitors as an antivirulence approach for Gram-negative pathogensMBio201456e0216525389178
  • GuckesKRKostakiotiMBrelandEJStrong cross-system interactions drive the activation of the QseB response regulator in the absence of its cognate sensorProc Natl Acad Sci U S A201311041165921659724062463
  • KostakiotiMHadjifrangiskouMCusumanoCKHannanTJJanetkaJWHultgrenSJDistinguishing the contribution of type 1 pili from that of other QseB-misregulated factors when QseC is absent during urinary tract infectionInfect Immun20128082826283422665375
  • NgWLPerezLCongJSemmelhackMFBasslerBLBroad spectrum pro-quorum-sensing molecules as inhibitors of virulence in vibriosPLoS Pathog201286e100276722761573
  • PerezLJKaragounisTKHurleyABasslerBLSemmelhackMFHighly potent, chemically stable quorum sensing agonists for Vibrio choleraeChem Sci20145115115524436778
  • HigginsDAPomianekMEKramlCMTaylorRKSemmelhackMFBasslerBLThe major Vibrio cholerae autoinducer and its role in virulence factor productionNature2007450717188388618004304
  • WeiYPerezLJNgWLSemmelhackMFBasslerBLMechanism of Vibrio cholerae autoinducer-1 biosynthesisACS Chem Biol20116435636521197957
  • TschowriNBusseSHenggeRThe BLUF-EAL protein YcgF acts as a direct anti-repressor in a blue-light response of Escherichia coliGenes Dev200923452253419240136
  • ObamaBExecutive order 13676: combating antibiotic-resistant bacteriaHouseTW184Washington, DCFederal Register20145693156935
  • National action plan for combating antibiotic-resistant bacteriaServices USDoHaHU.S. Government Publishing Office2015